EP1917276B1 - Procede de preparation d'interferon beta glycosyle - Google Patents

Procede de preparation d'interferon beta glycosyle Download PDF

Info

Publication number
EP1917276B1
EP1917276B1 EP05784517.4A EP05784517A EP1917276B1 EP 1917276 B1 EP1917276 B1 EP 1917276B1 EP 05784517 A EP05784517 A EP 05784517A EP 1917276 B1 EP1917276 B1 EP 1917276B1
Authority
EP
European Patent Office
Prior art keywords
ifn
cells
interferon beta
interferon
beta
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP05784517.4A
Other languages
German (de)
English (en)
Other versions
EP1917276A1 (fr
Inventor
Dina Fischer
Alain Bernard
Paul Ducommun
Mara Rossi
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ares Trading SA
Original Assignee
Ares Trading SA
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ares Trading SA filed Critical Ares Trading SA
Priority to RS20180470A priority Critical patent/RS57549B1/sr
Priority to EP20110161155 priority patent/EP2390263A1/fr
Priority to PT57845174T priority patent/PT1917276T/pt
Priority to PL05784517T priority patent/PL1917276T3/pl
Priority to SI200532202T priority patent/SI1917276T1/en
Publication of EP1917276A1 publication Critical patent/EP1917276A1/fr
Application granted granted Critical
Publication of EP1917276B1 publication Critical patent/EP1917276B1/fr
Priority to HRP20180579TT priority patent/HRP20180579T1/hr
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/0018Culture media for cell or tissue culture
    • C12N5/0031Serum-free culture media
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • A61K38/215IFN-beta
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/16Extraction; Separation; Purification by chromatography
    • C07K1/18Ion-exchange chromatography
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/16Extraction; Separation; Purification by chromatography
    • C07K1/20Partition-, reverse-phase or hydrophobic interaction chromatography
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • C07K14/565IFN-beta
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/0018Culture media for cell or tissue culture
    • C12N5/0037Serum-free medium, which may still contain naturally-sourced components
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/05Inorganic components
    • C12N2500/10Metals; Metal chelators
    • C12N2500/12Light metals, i.e. alkali, alkaline earth, Be, Al, Mg
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/32Amino acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/90Serum-free medium, which may still contain naturally-sourced components
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/90Serum-free medium, which may still contain naturally-sourced components
    • C12N2500/92Medium free of human- or animal-derived components
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/99Serum-free medium
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • C12N2510/02Cells for production

Definitions

  • the present invention relates to processes for producing recombinant human interferon beta under serum-free culture conditions.
  • Proteins have become commercially important as drugs that are also generally called "biologicals".
  • biologicals One of the greatest challenges is the development of cost effective and efficient processes for the production of recombinant proteins on a commercial scale.
  • Suitable cells that are widely used for production of polypeptides turned out to be Chinese Hamster Ovary (CHO) cells.
  • CHO cells were first cultured by Puck (1958) from a biopsy of an ovary from a female Chinese hamster. From these original cells a number of sub-lines were prepared with various characteristics.
  • One of these CHO cell lines, CHO-K1 is proline-requiring and is diploid for the dihydrofolate reductase (DHFR) gene.
  • Another cell line derived from this cell line is a DHFR deficient CHO cell line (CHO DUK B11) ( PNAS 77, 1980, 4216-4220 ), which is characterized by the loss of DHFR function as a consequence of a mutation in one DHFR gene and the subsequent loss of the other gene.
  • human cell lines such as the human fibrosarcoma cell line HT1080 or the human embryonic kidney cell line 293, a human embryonic retinoblast-derived cell line such as e.g. PER.C6, an amniotic cell derived-cell line or a neuronal-derived cell line.
  • Cells from a suitable cell line are stably transfected with an expression vector comprising the coding sequence of the protein of interest to be produced, together with regulatory sequences such as promoters, enhancers, or polyA signals that ensure stable and correct expression of the protein of interest.
  • Further genes usually present on expression vectors are marker genes such as e.g. positive selections markers (e.g. neo gene) that select the stably transfected cells from the untransfected and transiently transfected cells.
  • Amplifiable genes such as the DHFR gene are used for amplification of the coding sequences.
  • fed-batch or perfusion production mode is mainly dictated by the biology of the clone and the property of the product, and is done on a case-by-case basis during the course of the development of a new drug product (Kadouri and Spier 1997).
  • one of the culture systems of choice is stationary packed-bed bioreactor in which cells are immobilized onto solid carriers. This system is easy to operate and with appropriate carriers and culture conditions very high cell density (of ⁇ 10 7 -10 8 cell ⁇ ml -1 ) can be achieved.
  • serum serves as a universal nutrient for the growth and maintenance of all mammalian cell lines that produce biologically active products.
  • Serum contains hormones, growth factors, carrier proteins, attachment and spreading factors, nutrients, trace elements, etc.
  • Culture media usually contained up to about 10% of animal serum, such as fetal bovine serum (FBS), also called fetal calf serum (FCS).
  • FBS fetal bovine serum
  • FCS fetal calf serum
  • serum Although widely used, serum has many limitations. It contains high levels of numerous proteins interfering with the limited quantities of the desired protein of interest produced by the cells. These proteins derived from the serum must be separated from the product during downstream processing such as purification of the protein of interest, which complicates the process and increases the cost.
  • BSE Bovine Spongiform Encephalopathy
  • cell culture media comprise many components of different categories, such as amino acids, vitamins, salts, fatty acids, and further compounds:
  • EP 1 482 031 A describes a serum-free medium comprising HEPES, proline, and sodium chloride. However, it has never been disclosed or suggested to apply the medium for culturing a human interferon beta-producing cell.
  • Standard “basic media” are also known to the person skilled In the art. These media already contain several of the medium components mentioned above. Examples of such media that are widely applied are Dulbecco's Modified Eagle's Medium (DMEM), Roswell Park Memorial Institute Medium (RPMI), or Ham's medium.
  • DMEM Dulbecco's Modified Eagle's Medium
  • RPMI Roswell Park Memorial Institute Medium
  • Ham's medium Dulbecco's Modified Eagle's Medium
  • the cell culture harvest may e.g. be cell extracts for intracellular proteins, or cell culture supernatant for secreted proteins.
  • Ion exchange chromatographic systems have been used widely for separation of proteins primarily on the basis of differences in charge.
  • ion exchange chromatography charged patches on the surface of the solute are attracted by opposite charges attached to a chromatography matrix, provided the ionic strength of the surrounding buffer is low. Elution is generally achieved by increasing the ionic strength (i.e. conductivity) of the buffer to compete with the solute for the charged sites of the ion exchange matrix.
  • Changing the pH and thereby altering the charge of the solute is another way to achieve elution of the solute.
  • the change in conductivity or pH may be gradual (gradient elution) or stepwise (step elution).
  • Resins that may be used in ion exchange chromatography may contain different functional groups: diethylaminoethyl (DEAE) or diethyl-(2-hydroxypropyl)aminoethyl (QAE) have chloride as counter ion, while carboxymethyl (CM) and sulphopropyl (SP) have sodium as counter ion, for example.
  • DEAE diethylaminoethyl
  • QAE diethyl-(2-hydroxypropyl)aminoethyl
  • CM carboxymethyl
  • SP sulphopropyl
  • HIC hydrophobic interaction chromatography
  • RPLC reversed phase liquid chromatography
  • Reverse phase chromatography is a protein purification method closely related to HIC, as both are based upon interactions between solvent-accessible non-polar groups on the surface of biomolecules and hydrophobic ligands of the matrix.
  • ligands used in reverse phase chromatography are more highly substituted with hydrophobic ligands than HIC ligands. While the degree of substitution of HIC adsorbents may be in the range of 10-50 ⁇ moles/mL of matrix of C2-C8 aryl ligands, several hundred ⁇ moles/mL of matrix of C4-C8 alkyl ligands are usually used for reverse phase chromatography adsorbents.
  • the Source 30RPC column is a polymeric reverse phase matrix. It is based on rigid, monosized 30 micron diameter polystyrene/divinyl benzene beads. Its characteristics can be summarized as follows: Exceptionally wide pH range (1-12), high selectivity, high chemical resistance, high capacity and high resolution at high flow rates.
  • Size-exculsion chromatography also called gel-permeation chromatography (GPC)
  • GPC gel-permeation chromatography
  • Blue Sepharose is a chromatography resin based on a dye-ligand affinity matrix.
  • the ligand Cibacron Blue F3G-A, is covalently coupled to sepharoseTM through chlorotriazine ring (Clonis et al., 1987).
  • interferon beta is a naturally occurring soluble glycoprotein belonging to the class of cytokines.
  • interferons have a wide range of biological activities, such as anti-viral, anti-proliferative and immunomodulatory properties.
  • IFN-alpha The three major interferons are referred to as IFN-alpha, IFN-beta and IFN-gamma. These interferons were initially classified according to their cells of origin (leukocytes, fibroblasts or T-cells). However, it became clear that several types might be produced by one cell. Hence leukocyte interferon is now called IFN-alpha, fibroblast interferon is IFN-beta, and T-cell interferon is IFN-gamma. There is also a fourth type of interferon, lymphoblastoid IFN, produced in the "Namalwa" cell line (derived from Burkitt's lymphoma), which seems to produce a mixture of both leukocyte and fibroblast IFN.
  • Human fibroblast interferon has antiviral activity and is also known to inhi8bit proliferation of cells. It is a polypeptide of about 20,000 Da induced by viruses and double-stranded RNAs. From the nucleotide sequence of the gene for fibroblast interferon, cloned by recombinant DNA technology, Derynck et al., 1980 deduced the complete amino acid sequence of the protein, which is 166 amino acid long.
  • interferon- ⁇ has also been cloned.
  • US Patent No. 5,326,859 describes the DNA sequence of human IFN- ⁇ and a plasmid for its recombinant expression in bacteria such as E. coli.
  • European Patent No. 0 287 075 describes a CHO (Chinese Hamster Ovary) cell line, transfected with the interferon- ⁇ coding sequence and capable of producing recombinant interferon- ⁇ .
  • the protein is described as being glycosylated with a biantennary (two branched) oligosaccharide, featuring a single fucose moiety.
  • Interferon beta has been expressed in several cell lines, such as CHO cells, BHK 21 (baby hamster kidney cells) and LTK (mouse L-thymidine kinase negative) cells (Reiser and Hauser, 1987).
  • DHFR negative CHO cells have also been used for the expression of interferon beta (Innis and McCormick, 1982), (ChernaJovsky et al., 1984).
  • Interferons are known to be glycosylated, often with different glycoforms.
  • the saccharide structure of IFN- ⁇ was shown to include a bi-antennary structure, featuring a single fucose saccharide and terminal galactose sialylation (Conradt et al., 1987). Glycosylation was shown to also be important for solubility, since the IFN- ⁇ precipitated after deglycosylation with glycopeptidase F.
  • IFN- ⁇ produced by E. coli showed folding problems, due to lack of glycosylation in the bacterial expression system.
  • European Patent No. 0 529 300 describes a recombinant interferon- ⁇ having a specific glycosylation pattern, namely glycosylation with carbohydrate structures that feature one fucose per oligosaccharide unit. These carbohydrate structures are biantennary, triantennary and tetraantennary (two, three and four branched, respectively) oligosaccharides.
  • PCT Application No. WO 99/15193 also describes glycosylation of recombinant interferon- ⁇ featuring biantennary, triantennary and tetraantennary oligosaccharides.
  • the constituent monosaccharides included mannose, fucose, N-acetylglucosamine, galactose and sialic acid.
  • interferon beta is used as a therapeutic protein drug, a so-called biological, in a number of diseases, such as e.g. multiple sclerosis, cancer, or viral diseases such as e.g. SARS or hepatitis C virus infections.
  • diseases such as e.g. multiple sclerosis, cancer, or viral diseases such as e.g. SARS or hepatitis C virus infections.
  • the present invention is based on the development of a process for producing recombinant human interferon beta in a serum free medium.
  • the invention relates to a process for the manufacturing of glycosylated recombinant human interferon- ⁇ , comprising a step of culturing an interferon- ⁇ producing CHO cell in a serum-free medium, the serum-free medium comprising:
  • the present document further describes a process for purifying recombinant interferon beta from a fluid, in particular from the cell culture harvest derived from cells producing interferon beta.
  • the described process for the purification of recombinant human interferon from a fluid comprises the steps of:
  • interferon beta is a composition of differentially glycosylated interferon beta, I.e. an interferon beta having a unique glycosylation pattern or profile. Therefore, the present document also describes an interferon beta composition comprising an oligosaccharide structure comprising two or three fucose saccharides.
  • the first aspect of the present invention is based on the development of a process for the production of interferon beta under serum-free cell culture conditions.
  • the process for the manufacturing of glycosylated recombinant human interferon beta comprises a step of culturing an interferon beta producing CHO cell in a serum-free medium, the serum-free medium comprising:
  • the serum-free medium may e.g. comprise 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 21 mM of HEPES (2-(4-(2-HYDROXYETHYL)-1-PIPERAZINYL) ETHANESULFONIC ACID) buffer. It may also e.g. comprise 0.4, 0.5, 0.6,0.7, 0.8, 0.9, 1, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3, 3,1 mM of Proline.
  • Sodium chloride concentrations in the serum-free medium of the invention may e.g. be about 5400, 5500, 5600, 5700, 5800, 5900, 6000, 6100, 6200, 6300, 6400, 6500, 6600, 6700, 6800, 6900, 7000, 7100 mg/L.
  • the serum-free medium further comprises about 10 to about 20, preferably about 15 mg/L Phenol Red.
  • the Phenol Red concentration may e.g. be 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 mg/L.
  • the above-identified components may be used in any suitable known serum-free medium.
  • serum-free media are listed below: Medium Manufacturer Cat. No. EX-CELL 302 JRH 14312-1000M EX-CELL 325 JRH 14335-1000M CHO-CD3 Sigma C-1490 CHO III PFM Gibco 96-0334SA CHO-S-SFM II Gibco 12052-098 CHO-DHFR Sigma C-8862 ProCHO 5 Cambrex BE12-766Q SFM4CHO HyClone SH30549.01 Ultra CHO Cambrex 12-724Q HyQ PF CHO HyClone SH30220.01 HyQ SFX CHO HyClone SH30187.01 HyQ CDM4CHO HyClone SH30558.01 IS CHO-CD Irvine Scientific #91119 IS CHO-V Irvine Scientific #9197
  • the cell to be used in the process of the invention is an interferon beta expressing CHO clone such as e.g. the cell line described by Reiser and Hauser (1987) or the cells described by Innis and McCormick (1982).
  • interferon beta is also called IFN beta, or IFN- ⁇ , and encompasses interferon beta derived from any species and preferably human interferon beta, a 166 amino acid glycoprotein with a molecular weight of approximately 22,500 daltons.
  • interferon beta also encompasses functional derivatives, muteins, analogs, or fragments of IFN-beta.
  • interferon beta 1 a refers to glycosylated interferon beta.
  • interferon beta The activity of interferon beta may e.g. be measured using a reference standard calibrated against the World Health Organization natural interferon beta standard (Second International Standard for interferon, Human Fibroblast GB 23 902 531 ). The unit is expressed in international units (IU) of antiviral activity per mg of interferon beta-1a determined in an in vitro cytopathic effect bioassay using WISH cells and Vesicular Stomatitis virus. Conversion table for MIU and mcg of IFN-beta MIU 3 12 18 24 mcg 11 44 66 88
  • “Variants” or “muteins”, as used in the fame of the present invention, refer to analogs of IFN-beta, in which one or more of the amino acid residues of natural IFN-beta are replaced by different amino acid residues, or are deleted, or one or more amino acid residues are added to the natural sequence IFN-beta, without diminishing considerably the activity of the resulting products as compared with the wild type IFN-beta.
  • muteins are prepared by known synthesis and/or by site-directed mutagenesis techniques, or any other known technique suitable therefor.
  • variants or mutants in accordance with the present invention include proteins encoded by a nucleic acid, such as DNA or RNA, which hybridizes to DNA or RNA encoding IFN-beta as disclosed e.g. in US 4,738,931 under stringent conditions.
  • stringent conditions refers to hybridization and subsequent washing conditions, which those of ordinary skill in the art conventionally refer to as “stringent”. See Ausubel et al., Current Protocols in Molecular Biology, supra, Interscience, N.Y., ⁇ 6.3 and 6.4 (1987, 1992 ).
  • examples of stringent conditions include washing conditions 12-20°C below the calculated Tm of the hybrid under study in, e.g., 2 x SSC and 0.5% SDS for 5 minutes, 2 x SSC and 0.1% SDS for 15 minutes; 0.1 x SSC and 0.5% SDS at 37°C for 30-60 minutes and then, a 0.1 x SSC and 0.5% SDS at 68°C for 30-60 minutes.
  • stringency conditions also depend on the length of the DNA sequences, oligonucleotide probes (such as 10-40 bases) or mixed oligonucleotide probes. If mixed probes are used, it is preferable to use tetramethyl ammonium chloride (TMAC) instead of SSC. See Ausubel, supra.
  • Identity reflects a relationship between two or more polypeptide sequences or two or more polynucleotide sequences, determined by comparing the sequences. In general, identity refers to an exact nucleotide to nucleotide or amino acid to amino acid correspondence of the two polynucleotides or two polypeptide sequences, respectively, over the length of the sequences being compared.
  • a "% identity" may be determined.
  • the two sequences to be compared are aligned to give a maximum correlation between the sequences. This may include inserting "gaps" in either one or both sequences, to enhance the degree of alignment.
  • a % identity may be determined over the whole length of each of the sequences being compared (so-called global alignment), that is particularly suitable for sequences of the same or very similar length, or over shorter, defined lengths (so-called local alignment), that is more suitable for sequences of unequal length.
  • Any such variant or mutein preferably has a sequence of amino acids sufficiently duplicative of that of IFN-beta, such as to have substantially similar activity to IFN-beta.
  • a functional assay for evaluating whether any variant or mutein has a similar activity as IFN-beta is e.g. the assay measuring the activity of interferon on the cytopathic effect of vesicular stomatitis virus in WISH cells, e.g. described by Youcefi et al., 1985. Thus, it can be determined whether any given mutein has substantially the same activity as IFN-beta by means of routine experimentation.
  • Any such variant or mutein may have at least 40% identity or homology with the sequence of IFN-beta as disclosed e.g. in US 4,738,931 . More preferably, it has at least 50%, at least 60%, at least 70%, at least 80% or, most preferably, at least 90% identity or homology thereto.
  • Muteins of IFN-beta which can be used in accordance with the present invention, or nucleic acid coding therefor, include a finite set of substantially corresponding sequences as substitution peptides or polynucleotides which can be routinely obtained by one of ordinary skill in the art, without undue experimentation, based on the teachings and guidance presented herein.
  • amino acids may include synonymous amino acids within a group which have sufficiently similar physicochemical properties that substitution between members of the group will preserve the biological function of the molecule (Grantham, 1974). It is clear that insertions and deletions of amino acids may also be made in the above-defined sequences without altering their function, particularly if the insertions or deletions only involve a few amino acids, e.g., under thirty, and preferably under ten, and do not remove or displace amino acids which are critical to a functional conformation, e.g., cysteine residues. Proteins and muteins produced by such deletions and/or insertions come within the purview of the present invention.
  • Examples for of amino acid substitutions in proteins which can be used for obtaining muteins of IFN-beta for use in the present invention include any known method steps, such as presented in US patents 4,959,314 , 4,588,585 and 4,737,462, to Mark et al ; 5,116,943 to Koths et al. , 4,965,195 to Namen et al ; 4,879,111 to Chong et al ; and 5,017,691 to Lee et al ; and lysine substituted proteins presented in US patent No. 4,904,584 (Shaw et al ).
  • Consensus interferons are non-naturally occurring variants of IFN ( US 6,013,253 ). Consensus interferons may also be produced according to the invention.
  • “Functional derivatives" of IFN-beta as used herein covers derivatives which may be prepared from the functional groups which occur as side chains on the residues or the N- or C-terminal groups, by means known in the art, and are included in the invention as long as they remain pharmaceutically acceptable, i.e., they do not destroy the biological activity of the proteins as described above, i.e., the ability to bind the corresponding receptor and initiate receptor signaling, and do not confer toxic properties on compositions containing it.
  • Derivatives may have chemical moieties, such as carbohydrate or phosphate residues, provided such a derivative retains the biological activity of the protein and remains pharmaceutically acceptable.
  • interferon beta may, for example, include polyethylene glycol side-chains, which may improve other properties of the protein, such as the stability, half-life, bioavailability, tolerance by the human body, or immunogenicity.
  • IFN-beta may be linked e.g. to Polyethlyenglycol (PEG). PEGylation may be carried out by known methods, described in WO 92/13095 , for example.
  • PEG-IFN can be prepared in accordance with the teaching of WO 99/55377 .
  • a functional derivative of IFN-beta may comprise at least one moiety attached to one or more functional groups, which occur as one or more side chains on the amino acid residues.
  • An embodiment in which the moiety is a polyethylene glycol (PEG) moiety is highly preferred.
  • PEG moieties may also be attached to the IFN-beta.
  • Other derivatives include aliphatic esters of the carboxyl groups, amides of the carboxyl groups by reaction with ammonia or with primary or secondary amines, N-acyl derivatives of free amino groups of the amino acid residues formed with acyl moieties (e.g. alkanoyl or carbocyclic aroyl groups) or O-acyl derivatives of free hydroxyl groups (for example that of seryl or threonyl residues) formed with acyl moieties.
  • acyl moieties e.g. alkanoyl or carbocyclic aroyl groups
  • O-acyl derivatives of free hydroxyl groups for example that of seryl or threonyl residues
  • a “fragment” according to the present invention refers to any subset of IFN-beta, that is, a shorter peptide, which retains the desired biological activity as measurable e.g. in the bioassay described above. Fragments may readily be prepared by removing amino acids from either end of the molecule and testing the resultant for its properties as a receptor agonist. Proteases for removing one amino acid at a time from either the N-terminal or the C-terminal of a polypeptide are known, and so determining fragments, which retain the desired biological activity, may be determined e.g. in the test described by Youcefi et al., 1985, and involves only routine experimentation.
  • the process for production of interferon beta of the invention comprises a growth phase I, a growth phase II and a production phase, wherein the growth phase I is carried out at 37 °C, the growth phase II is carried out at 35 °C, and the production phase is carried out at 33 °C.
  • growth phase I may e.g. be 10 to 12 days.
  • Growth phase II is generally shorter and mainly serves for adapting the cells to a lower temperature. According to the invention, the growth phase II takes 1 to 2 days.
  • the process of the invention may be carried out as a fed-batch or a perfusion process.
  • perfusion is preferred.
  • the process is a perfusion process with a dilution rate ranging from about 1 to about 10, preferably from about 1.5 to about 7 per day.
  • the dilution rate may be in a range of e.g. 0.5, 1, 1.5, 1.6, 1.7, 1.8, 1.9, 2, 2.5, 3, 3.5,4, 4.5, 5, 5.5, 6, 6.5, 7, 7.1, 7.2, 7.3, 7.4, 7.5, 8, 8.5, 9, 9.5, 10, 10. 5.
  • the dilution rate is increased within the first two to three weeks of cell culture from an initial value of about 1 to 2 per day to a value of about 7 to 10 per day, particularly during the production phase.
  • the culturing step of the process of the invention may be carried out in any suitable environment, such as Petri dishes, T-flasks or roller bottles, but preferably in vessels having greater volumes such as e.g. a bioreactor.
  • the system may e.g. be a stationary packed-bed bioreactor in which cells are immobilized onto solid carriers.
  • This system is easy to operate and with appropriate carriers and culture conditions very high cell density (of ⁇ 10 7 -10 8 call ⁇ ml -1 ) can be achieved.
  • a solid carrier that may be used in accordance with the present invention may e.g. be a microcarrier.
  • Microcarriers are small solid particles on which cells may be grown in suspension culture. Cells are capable of adhering and propagating on the surface of microcarriers. Typically, microcarriers consist of beads, the diameter of which is comprised between 90 ⁇ m and 300 ⁇ m. Microcarriers can be made of various materials that have proven successful for cell attachment and propagation such as, e.g., glass, polystyrene, polyethylene, dextran, gelatin and cellulose.
  • microcarriers may be coated with a material promoting cell attachment and growth such as, e.g., e.g., N,N-diethylaminoethyl, glass, collagen or recombinant proteins.
  • a material promoting cell attachment and growth such as, e.g., e.g., N,N-diethylaminoethyl, glass, collagen or recombinant proteins.
  • Both macroporous and non-porous microcarriers do exist. Macroporous surfaces give the cells easy access to the interior of the microcarrier after inoculation, and once inside of the microcarrier, the cells are protected from the shear forces generated by mechanical agitation and aeration in the bioreactor.
  • a further solid carrier that may be used in accordance with the present invention may e.g. be a disk, such as a disk composed of polyester non-woven fiber bonded to a sheet of polypropylene mesh (see, e.g., U.S. patent No. 5,266,476 ).
  • Such disks are usually treated electrostatically to facilitate suspension cells adhering to the disks and becoming trapped in the fiber system, where they remain throughout the cultivation process. Cell density and productivity achieved with cells grown on disks can be up to ten times higher than with cells growing on microcarriers.
  • the process for the production of glycosylated interferon beta preferably further comprises a step of collecting the interferon beta containing cell culture harvest.
  • the cell culture harvest is further subjected to a purification process.
  • the purification process may be any process leading to interferon beta of the required purity and may contain any combination of purification steps based on chromatography or any other purification technology such as fractionation with salt or the like.
  • the purification is preferably carried out by a process for the purification of recombinant interferon from a fluid, comprising the steps of:
  • Step (a) is preferably carried out on Blue Sepharose, e.g. on a Blue Sepharose fast flow column.
  • Step (b) is preferably carried out on a caboxymethyl resin, e.g. on a CM Sepharose fast flow.
  • the purification process may further comprise , before step (a), a step of clarifying the fluid by microfiltration.
  • the purification process may further comprise the steps of:
  • Step (f) may be carried out e.g. by micro- or nanofiltration.
  • Ultrafiltration is useful for removal of small molecular weight components in the eluates resulting from previous chromatrographic steps. Ultrafiltration e.g. allows to remove organic solvent, TFA and salts from the previous step, to equilibrate the interferon beta In the required buffer, or to concentrate the molecule to the desired concentration. Such ultrafiltration may e.g. be performed on ultrafiltration media excluding components having molecular weights below 5 kDa.
  • a virus removal filtration step may e.g. be carried out between steps (d) and (e), or after step (e). More preferably, the process comprises two virus removal steps.
  • the purity that may be obtained with the purification process according to the invention is preferably > 80%, more preferably > 90 % and most preferably > 98%.
  • the process of purifying Interferon beta preferably further comprises a step of formulating the purified interferon beta into a pharmaceutical composition, optionally together with a pharmaceutically acceptable carrier.
  • a chinese hamster ovary (CHO) cell line designated DUKX-B11, which lacks DHFR (dihydrofolate reductase) activity, is preferably used as the host cell for the preparation of glycosylated interferon beta.
  • DUKX-B11 which lacks DHFR (dihydrofolate reductase) activity
  • DHFR dihydrofolate reductase
  • the DNA sequence coding human interferon- ⁇ is e.g. described in US patent no. 5,326,859 .
  • a preferred embodiment of the present invention relates to a method for producing recombinant human interferon- ⁇ in transfected cells capable of producing at least about 100000 IU of recombinant human interferon- ⁇ in specific cellular productivity (IU/10 6 cells/24 hours).
  • the cells are capable of producing at least about 200000 IU or at least about 200000 IU or at least about 300000 IU or at least about 400000 IU or at least about 500000 IU or at least about 600000 IU of recombinant human interferon beta specific cellular productivity.
  • the interferon beta producing cell is a CHO cell which is transfected with a nucleic acid construct comprising at least one promoter/enhancer element functionally linked to the human IFN- ⁇ gene. More preferably, the at least one promoter/enhancer element comprises a SV40 promoter/enhancer. Most preferably, the nucleic acid construct comprises at least a first transcription unit composed of the SV40 promoter/enhancer functionally linked to the human IFN- ⁇ gene, the human IFN- ⁇ gene being functionally linked to the SV40 T Ag early polyadenylation region. It is also highly preferred that the nucleic acid construct further comprises at least a second transcription unit composed of a SV40 promoter/enhancer, a mouse DHFR gene and a SV40 T Ag polyA-containing early polyadenylation region.
  • the present document also describes a nucleic acid construct comprising at least one promoter/enhancer element functionally linked to the human IFN- ⁇ gene for being transfected into cells, being characterized in that the transfected cells are capable of producing at least about 100000 IU, at least about 200000 IU or at least about 300000 IU or at least about 400000 IU or at least about 500000 IU or at least about 600000 IU of recombinant human interferon- ⁇ in specific cellular productivity (IU/10 6 cells / per 24 hours).
  • At least one promoter/enhancer element comprises a SV40 promoter/enhancer.
  • the nucleic acid construct comprises at least a first transcription unit composed of the SV40 promoter/enhancer functionally linked to the human IFN- ⁇ gene, the human IFN- ⁇ gene being functionally linked to the SV40 T Ag early polyadenylation region.
  • the nucleic acid construct further comprises at least a second transcription unit composed of a SV40 promoter/enhancer, a mouse DHFR gene and a SV40 T Ag polyA-containing early polyadenylation region.
  • the document further describes an interferon beta obtainable by a process according to the present invention.
  • interferon beta composition having a unique glycosylation profile.
  • interferon beta is preferably produced by a process according to the present invention.
  • the glycosylated recombinant human interferon- ⁇ protein may contain an oligosaccharide structure having two or three fucose saccharides.
  • the oligosaccharide structure further comprises a disialyl biantennary trifucosylated glycan (Neu 2 Ac.Hex 5 .HexNAc 4 .Fuc 3 ).
  • the unique glycosylation pattern further comprises a non sialylated biantennary structure (Hex 5 .HexNAc 4 .Fuc); disialylated triantennary structure or disialylated biantennary with N-acetyl lactosamine repeat structures (NeuAc2.Hex6.HexNAc5.Fuc); trisialylated triantennary structure (NeuAc 3 .Hex6.HexNAc 5 .Fuc); trisialylated triantennary structure with N-acetyl lactosamine repeat structures or trisialylated tetrantennary (NeuAc 3 .Hex 7 .HexNAc 6 .Fuc); mono sialylated and disialylated biantennary structure with two fucose units (NeuAc.Hex 5 .HexNAc 4 .Fuc 2 ,NeuAc 2 ,NeuAc 2
  • the unique glycosylation pattern comprises glycans featuring a similar level of N-Acetylneuraminic acid : N-Glycolylneuraminic acid as for natural human protein glycosylation patterns.
  • the interferon beta composition is characterized by a sialylation profile comprising about 1 to about 5% of unsialylated N-glycans, about 5 to about 25% of mono-sialylated glycans, about 55 to about 75% of disialylated N-glycans, about 10 to about 25% of tri-sialylated N-glcyans.
  • interferon beta for the manufacture of a medicament for treatment of human disease, in particular multiple sclerosis, cancer, or viral infections.
  • the multiple sclerosis may be selected from the group consisting of relapsing, non-relapsing and early onset multiple sclerosis.
  • a method of treating a subject in need of treatment with interferon- ⁇ may comprise administering to the subject recombinant human interferon- ⁇ protein as described herein.
  • the treatment is for multiple sclerosis. More specifically, the multiple sclerosis may be selected from the group consisting of relapsing, non-relapsing and early onset multiple sclerosis.
  • the treatment is an anti-tumor treatment.
  • the treatment is an anti-viral treatment.
  • the present document further describes a pharmaceutical composition
  • a pharmaceutical composition comprising, as an active ingredient, an isolated, purified, recombinant human interferon- ⁇ composition as described herein and a pharmaceutically acceptable carrier for administration of the pharmaceutical composition.
  • the pharmaceutical composition having anti-tumor or anti-viral activity, or activity against multiple sclerosis comprises, as an active ingredient, an isolated, purified, recombinant human interferon- ⁇ protein as described herein and a pharmaceutically acceptable carrier for administration of the pharmaceutical composition.
  • an article of manufacture comprising packaging material and a therapeutically effective amount of an isolated, purified recombinant interferon- ⁇ protein, wherein the packaging material comprises a label or package insert indicating that the recombinant human interferon- ⁇ protein as described herein can be administered to a human for treatment thereof.
  • the isolated, purified, recombinant human interferon- ⁇ protein or pharmaceutical composition as described herein is preferably used for the manufacture of a medicament for treatment of relapsing, non-relapsing and early onset multiple sclerosis.
  • Dosing regimens for a particular subject (patient) can easily be determined by one of ordinary skill in the art, as these regimens are well known in the art.
  • This Example describes the generation of an interferon beta producing CHO clone.
  • FIG. 1 An overview over the process of generating the clone is depicted in Fig. 1 .
  • a DNA fragment comprising the human interferon- ⁇ coding region was isolated from a human peripheral blood cell genomic DNA library.
  • a DHFR-deficient CHO cell line was transfected with a recombinant plasmid containing both the human IFN- ⁇ coding sequence and the mouse DHFR gene as a selectable and amplifiable marker. After selection in thymidine-free medium, gene amplification with methotrexate (MTX), and cloning, a cell producing IFN- ⁇ -1a at high levels was isolated. The cells were subjected to genotypic and phenotypic characterization.
  • MTX methotrexate
  • the expression vector containing the IFN- ⁇ coding sequence was devoid of the IFN- ⁇ 3'UTR and thus of the IFN- ⁇ polyadenylation region.
  • the expression vector therefore contained two transcription units, a first IFN- ⁇ transcription unit composed of the SV40 promoter/enhancer, the human IFN- ⁇ coding sequence and the SV40 T Ag early polyadenylation region, and a second DHFR transcription unit composed of the SV40 promoter/enhancer, the mouse DHFR gene and the SV40 TAg polyA-containing early polyadenylation region.
  • the structure of the expression vector was verified by restriction map analysis and by complete sequencing (double-stranded, automated sequencing). The correct sequence of the fragments used for Its construction was confirmed in both directions.
  • the cell line was isolated from the CHO-K1 cell line, requiring proline (Kao and Puck, 1968) by mutagenesis with ethyl methanesulfate followed by gamma Irradiation.
  • DHFR deficient mutants were selected by exposure to high specific activity [ 3 H] - deoxyuridine (Urlaub and Chasin, 1980).
  • DHFR-deficient mutants require glycine, hypoxanthine and thymidine for growth.
  • anchorage-dependent, DHFR-deficient CHO cells were transfected by the calcium phosphate precipitation procedure (Graham FL and Van Der EB, 1973; Busslinger, et al., 1981) with the plasmid containing both the h-IFN- ⁇ coding sequence and the mDHFR marker gene described above.
  • MTX metalhotrexate
  • the CHO DHFR-deficient cell line DUKX-B11 was cultured in Ham's Nutrient Mixture F12 supplemented with 10% FBS, at 37°C, 5% CO 2 .
  • CaPO 4 -DNA co-precipitates were prepared by mixing the vector DNA, dissolved in 0.45 ml of 10mM Tris-HCl pH 7.9, 0.1 mM EDTA, with 0.05 ml of a 2.5M CaCl 2 solution.
  • the cells were then trypsinized and subcultured at 1:4 to 1:10 split ratio in selective medium consisting of Dulbecco's Modified Eagles Medium (DMEM) lacking thymidine but supplemented with 150 ⁇ g/ml proline and 10% dialyzed FBS.
  • DMEM Dulbecco's Modified Eagles Medium
  • the cultures were kept at 37°C and 8% CO 2 and the selective medium was changed every 3-4 days.
  • Interferon beta producing cells were isolated after 10-12 days by trypsinization with 3mm trypsin-soaked paper-discs. Forty-three clones were picked, individual clones were grown and the cell culture supernatants tested for hIFN- ⁇ production by ELISA. Three clones producing more than 30,000 IFN- ⁇ IU/10 6 cells/24 hours were selected for gene amplification.
  • Each of the clones was subjected to culture with low concentrations of MTX. The entire cell population that survived the treatment was subjected to higher MTX concentrations. Subcloning and clone selection were performed only after the last amplification stage. The selected high producers (more than 400,000 IU/10 6 cells /24 hours) were subjected to clone stability studies. A relatively stable high producer clone was selected and subcloned. From the resulting clones, a high producer stable clone was selected.
  • Amplification increased production levels (specific productivity) of IFN- ⁇ -1a from 30,000 to 500,000 IU/10 6 cells/24 hours, as determined by ELISA.
  • the blot was prepared as follows.
  • tissue culture (growth) medium was DMEM (Dulbecco's Modified Eagle's medium), supplemented with proline (150mg/l) and 10% FBS (fetal bovine serum) or in serum-free medium, e.g. Ex-Cell 302 from JRH.
  • DMEM Dulbecco's Modified Eagle's medium
  • proline 150mg/l
  • FBS fetal bovine serum
  • Cells from the interferon producing clone were seeded in TC80 flasks (2 x 10 6 cells/flask) in 30 ml growth medium (either DMEM and FBS, or serum-free medium). When initial confluency was reached, as determined by microscopic examination, the growth medium was replaced with 20 ml of fresh medium and the cultures incubated at about 32°C for 24 hours. Samples of the culture medium were obtained from each flask. IFN- ⁇ -1a level was determined by ELISA from the culture medium with a commercial ELISA kit (for example, the Toray ELISA kit from Toray, Japan).
  • a commercial ELISA kit for example, the Toray ELISA kit from Toray, Japan.
  • Specific cellular productivity was calculated by multiplying the IU/ml of IFN- ⁇ -1a produced per 24 hours by the volume per TC80 flask and dividing by the total cell number (in millions) per flask.
  • the interferon beta producing cell clone was a stable cell line, having a high production capacity for recombinant human interferon- ⁇ in the range of about 100,000 IU in specific cellular productivity (IU/10 6 cells / 24 hours) and about 600,000 IU in specific cellular productivity.
  • the mean productivity of the new cell line was 556,000 ⁇ 119,000 IU/10 6 cells/24 hrs.
  • PCR DNA products derived from the hIFN- ⁇ messenger RNA were used to determine the coding region nucleotide sequences as the mRNA sequence provides direct proof that the RNA transcripts are processed correctly.
  • RNA Total cellular RNA was prepared from the cells in the exponential stage of growth in T-flask cultures (Chomczynski and Sacchi, 1987).
  • Complementary DNA cDNA was synthesized from the mRNA samples in a reaction which contained 2 micrograms ( ⁇ g) of total RNA, 0.5 ⁇ M random hexamers, 2.5 mM 1X PCR II buffer [10 mM Tris-HCl (pH 8.3), 50 mM KCI], 0.5 mM each of dATP, dCTP, dGTP, and dTTP, 40 units of RNase Inhibitor, and 200 units of Reverse Transcriptase in a final volume of 100 ⁇ l.
  • cDNA Complementary DNA
  • RNA template, primer and nuclease-free water were combined and incubated at 65°C for 10 minutes and were then placed in an ice bath. The remaining components were added and the reaction was incubated at 42°C for 60 minutes, then 70°C for 15 minutes and was then held at 4°C indefinitely.
  • the RT products (cDNAs) were stored at - 20°C until further use. As a control, a reaction with all components except the reverse transcriptase was prepared. The "no RT" control is to exclude the unlikely possibility that the RNA preparations had DNA contamination.
  • PCR amplification was done using primers SRB1 AP1 and AP2 for the cDNA templates. Sequences for these primers are as follows:
  • the PCR reactions consisted of the following: 4.0 ⁇ l of cDNA reaction mixture, 50 pmol of each of the primer pair, and 25 ⁇ l HotStarTaqTM Master Mix in a reaction volume of 50 ⁇ l.
  • the reactions were heated at 95°C for 15 minutes followed by 30-35 cycles of: (a) 94°C for 30 seconds, (b) 55°C for 30 seconds, and (c) 72°C for 1 minute.
  • the hIFN- ⁇ PCR products were purified by low melting point (LMP) agarose gel electrophoresis followed by extraction using the QIAquick gel extraction kit (Qiagen).
  • PCR products were sequenced directly with the Big DyeTM Terminator Cycle Sequencing Ready Reaction Kit with AmpliTaq® DNA Polymerase. All sequencing reactions were analyzed on 5.75% Long RangerTM gels on ABI373-S automated DNA sequencers. The raw data were tracked and analyzed using ABD Analysis software.
  • PCR amplification resulted in the generation of the predicted approximate 815 bp fragment. No PCR products were observed for the "no RT" control nor for the other negative controls.
  • the sequencing data demonstrate that the hIFN- ⁇ gene integrated into cells of genome of the clone is correctly transcribed into hIFN- ⁇ mRNA.
  • the gene copy number was determined by Southern blot analysis of BamHI digests.
  • the specific primers used to build the probes for the gene copy number analysis are the following:
  • Genomic DNA was isolated from exponentially growing T-flask cultures of the cells using a modification of the salting out method (Martinez et al., 1998). Briefly, the cells were resuspended in a Tris-NaCl-EDTA buffer and then lysed with a Tris-NaCl-EDTA-SDS buffer. This suspension was treated overnight with proteinase K. After addition of a saturated salt solution and centrifugation, the genomic DNA was precipitated by addition of isopropanol to the aqueous phase. Following a 70% ethanol wash, the DNA pellet was resuspended in a TE/RNase A solution (10 mM Tris-HCl pH 8.0, 1 mM EDTA, 20 ⁇ g/ml RNaseA).
  • Size determinations were made based on the migration of the bands as visualized by 32 P on an autoradiograph.
  • the expression vector was used as the control.
  • Copy number determinations were based on relative 32 P levels in the bands as quantitated on a model 445SI PhosphoImagerTM (Molecular Dynamics; Sunnyvale, CA). Autoradiographs were photographed to provide a record of the results (data not shown).
  • Enzyme digestion of the DNA extracted from the interferon beta producing cell line resulted in the production of prominent bands that matched those predicted from the expression vector for the BamHI, BglI, Dral and HincII digests.
  • a very faint band was observed with the Bgll-digested genomic DNAs, most likely a result of incomplete digestion.
  • restriction enzymes flank the hIFN- ⁇ expression unit indicating that an intact, functional, full-length unit has integrated into the genome.
  • the probes used included a 32 P-labeled hIFN- ⁇ probe prepared as described in section "Determination of gene copy number" and a control G3PDH cDNA probe (Clontech; Palo Alto, CA).
  • RNA Total RNA, 5 ⁇ g per lane, was size-fractionated by electrophoresis in agarose gels that contained formaldehyde as a denaturant. Samples were loaded in duplicate sets. The RNA was transferred in 10X SSC to nylon membranes by capillary action. Prehybridization and hybridization were done at 65°C in the modified Church and Gilbert solution described in section "Restriction endonuclease map analysis". The blots were hybridized to a 32 P-labeled hIFN- ⁇ probe and a control G3PDH cDNA probe. The band sizes were estimated from an autoradiograph of the blots.
  • the mRNA size was estimated to be 0.9 kb mRNA. This size correlates well to an mRNA starting at the SV40 transcription initiation site and resulting in a transcript of about 800 nucleotides without considering the polyA tail.
  • the phenotypic and genotypic studies on the interferon beta producing cells confirmed the identity and consistency of the cells.
  • the chromosomal integration of the hIFN- ⁇ gene into the CHO cells genome was demonstrated by in-situ hybridization studies.
  • the hIFN- ⁇ gene copy number levels were measured by Southern blot analysis of DNA extracted from the cells of the described clone. The results showed that gene copy number levels were in the same ranges for all cells, independently of the population doubling levels.
  • RNA prepared from the cells showed a single band of approximately 0.9 kb in size.
  • interferon beta producing cells synthesized hIFN- ⁇ mRNA transcripts with the correct protein coding region sequence, indicating that the interferon beta producing cells produce human recombinant IFN- ⁇ (IFN- ⁇ -1a) with the correct primary amino acid sequence.
  • restriction map analyses were performed on the interferon beta producing cells. Multiple digests were separated by agarose gel electrophoresis, transferred to nylon membranes and hybridized to labelled probes specific for the hIFN- ⁇ . Consistent restriction fragment profiles, Indicated the integration of an intact functional hIFN- ⁇ expression unit, in all the cell banks.
  • the hIFN- ⁇ gene copy numbers were determined by Southern blot analysis of DNA extracted from the interferon beta producing cells (data not shown). The results show gene copy numbers to be about 100 copies per cell, which is about four times higher than for the clone described in the literature (Chernajovsky et al., 1984).
  • the hIFN- ⁇ cDNAs prepared from the cells' mRNAs were sequenced and the results showed that for the cells, the hIFN- ⁇ gene sequence was 100% Identical to the expected sequences. Thus, the hIFN- ⁇ gene is correctly transcribed into mRNA.
  • the genomic DNA sequence of the 5' and 3' regions flanking the hIFN- ⁇ gene was determined for the cells of the clone and found to be 100% identical to the corresponding sequences of expression vector and the published sequence of the hIFN- ⁇ gene.
  • transfected gene is stably integrated in the interferon beta producing cells' genome.
  • the overall goal of this experiment was to develop a process for producing IFN beta-1a from the clone described in example 1 under serum-free conditions.
  • the serum-free process was developed at 75L bioreactor scale with internal packed bed Fibra-Cel® carriers.
  • the cells were thawed and expanded over 21 days in a commercially available serum-free medium having the following modifications: Table 1 Modification of serum-free medium Ingredients Composition in % (w/w) HEPES 20 mM Proline 1 mM Phenol Red 15 mg/L Sodium chloride 6150 mg/L 30 ⁇ 10 9 cells were seeded in the 75L bioreactor (high seeding).
  • This temperature strategy resulted in a productivity around 6.0 x 10 6 IU /ml bio per day. This productivity is about five times higher than the productivity of the clone described in the literature (Chemajovsky et al., 1984) in serum-containing medium.
  • NAC N-Actey-Cyteine
  • Zinc Zinc
  • a 66% lower seeding cell density (1.3.10 9 cells) was also tested in order to evaluate its impact on growth phase duration, metabolism and productivity.
  • the purification process of the IFN- ⁇ -1a from cell culture supernatant included four chromatographic and four filtration stages, as shown in Fig. 2 .
  • the purification stages were performed in the following order:
  • the purification of the active ingredient started with dye affinity chromatography on Blue Sepharose 6 FF (BS 6 FF), which was the major purification stage.
  • BS 6 FF Blue Sepharose 6 FF
  • the amount of host cell derived proteins as well as DNA from ruptured CHO cells was reduced by several orders of magnitude and therefore IFN- ⁇ -1a in the BS 6 FF eluate was significantly enriched.
  • ultrafiltration was performed to reduce the solution volume and exclude low molecular weight material.
  • IFN- ⁇ -1a three main types of column-based protein separations have been chosen. Ion-exchange chromatography on CM-sepharose FF resin removes nucleic acid and FBS/CHO derived proteins. Reverse phase HPLC reduced pyrogens, residual host cell derived proteins and degraded forms of IFN- ⁇ -1a. A final polishing stage of gel filtration was performed using Sephacryl S-100 HR resin. The eluate was microfiltered (0.22 ⁇ m) and stored at -70°C or below.
  • glycosylated proteins often occur as a mixture of different glycoforms, or proteins having different saccharide structures in their glycosylation.
  • Various techniques were used to analyze these glycoforms, as described in greater detail below, including electrospray mass spectroscopy, FAB-MS, MALDI-MS, tandem mass spectroscopy (MS/MS) and GC-MS (linkage studies). These different techniques all showed that of the different saccharide structures studied, one such structure was newly present in the IFN- ⁇ -1a obtained from the clone.
  • ES-MS electrospray mass spectrometry
  • MS/MS of glycans This technique permitted rapid monitoring of the glycoform distribution of the bulk samples at the molecular mass level. This method is e.g. described by Domon, B. and Costello, CE (1988).
  • the major glycoforms were the core-fucosylated disialyl (peak C) and monosialyl (peak B) biantennary carbohydrate structures and the minor glycoforms corresponded to the core-fucosylated non-sialylated biantennary (peak A), the core-fucosylated triantennary trisialylated (peak E) and 2 other core-fucosylated minor complex structures (peaks D and F).
  • the glycoform pattern of IFN- ⁇ -1a derived from interferon beta obtained by the new process was analyzed by ES-MS.
  • IFN- ⁇ -1a bulk samples derived from interferon beta obtained by the new process were subjected to extensive carbohydrate characterization studies.
  • the carbohydrate composition of IFN- ⁇ -1a was obtained using FAB-MS, MALDI-MS, Nanospray-MS/MS analyses and linkage studies (GC-MS) of permethylated IFN- ⁇ -1a following tryptic and peptide N-glycosidase F digestion.
  • Glycosylation site determination was accomplished by FAB-MS analyses of chymotryptic peptides previously digested with trypsin and N-glycosidase F.
  • the tryptically cleaved peptide glycopeptide mixture from the IFN- ⁇ -1a was treated with the enzyme peptide-N-glycosidase F (e.g. as described by Tarentino et al.1985).
  • a portion of the permethylated glycan was analyzed by positive ion FAB-MS (obtained in low mass ranges for fragment ions and high mass ranges for molecular ions), as described e.g. by Barber, et al. (1981) and Taylor (1983).
  • MALDI- mass spectrometry was performed as e.g. described by Hillenkamp, et al., 1991.
  • Nanospray-mass spectrometry was performed as described e.g. by Wilm and Mann 1996.
  • the tryptic peptides were digested with peptide N-glycosidase F and purified by Sep-pak.
  • the propanol fractions (20%-40%) of the Sep-pak were sub-digested with chymotrypsin and analysed by FAB/MS.
  • the relative abundance of N-glycolylneuraminic acid was calculated from the peak heights of signals in the permethylated N-linked glycan FAB and MALDI-TOF data.
  • the IFN- ⁇ -1a glycoforms contain mainly N-Acetylneuraminic acid like most human glycoproteins.
  • Figure 6 shows the MALDI spectrum of IFN- ⁇ -1a with permethylated glycans (list of signals in Table 3).
  • the IFN- ⁇ -1a obtained from the process according to the present invention contains the trifucose structure, NeuAc 2 .Hex 5 .HexNAc 4 .Fuc 3 as previously described.
  • Nanospray MS/MS of the permethylated glycans confirmed the detection of trace levels of NeuAc 2 .Hex 5 .HexNAc 4 .Fuc 2 oligosaccharides in all samples while traces of NeuAc 2 .Hex 5 .HexNAc 4 .Fuc 3 were observed only in IFN- ⁇ -1a from the process of the present invention.
  • the relative abundance of N-glycolylneuraminic acid was calculated from the peak heights of signals in the permethylated N-linked glycan FAB and MALDI-MS data.
  • N-acetylneuraminic acid:N-glycolylneuraminic acid ratio was 31.7:1.0 in a sample and suggest that 3.1% of the sialic acid is N-Glycolylneuraminic acid. These results are in agreement with similar levels (5 %) obtained for natural human interferon produced by human fibroblasts. As expected the IFN- ⁇ -1a glycoforms contain mainly N-acetylneuraminic acid like most human glycoproteins.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Biotechnology (AREA)
  • Wood Science & Technology (AREA)
  • Biochemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Cell Biology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Toxicology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Analytical Chemistry (AREA)
  • Immunology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Epidemiology (AREA)
  • Virology (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Hospice & Palliative Care (AREA)

Claims (5)

  1. Procédé pour la fabrication d'interféron bêta humain recombinant glycosylé, comprenant une étape de culture d'une cellule CHO productrice d'interféron bêta humain, dans un milieu sans sérum, le milieu sans sérum comprenant :
    - 10 à 30 mM d'HEPES, de préférence 20 mM d'HEPES ;
    - 0,5 à 3 mM de proline, de préférence 1 mM de proline ; et
    - 5 500 à 7 000 mg/l de chlorure de sodium, de préférence 6 100 mg/l de chlorure de sodium,
    le procédé comprenant une phase de croissance I, une phase de croissance II et une phase de production, la phase de croissance I étant effectuée à 37 °C, la phase de croissance II étant effectuée à 35 °C et la phase de production étant effectuée à 33 °C et la phase de croissance II durant 1-2 jours.
  2. Procédé selon la revendication 1, le milieu sans sérum comprenant 10 à 20 mg/l de rouge de phénol, de préférence 15 mg/l de rouge de phénol.
  3. Procédé selon l'une quelconque des revendications précédentes, le procédé étant un procédé de perfusion avec un taux de dilution valant de 1 à 10, de préférence de 1,5 à 7 par jour.
  4. Procédé selon la revendication 3, dans lequel on augmente le taux de dilution pendant les deux à troisième premières semaines de culture des cellules, en partant d'une valeur initiale de 1 à 2 par jour, jusqu'à une valeur de 7 à 10 par jour.
  5. Utilisation d'un milieu de culture sans sérum, comprenant :
    - 10 à 30 mM d'HEPES, de préférence 20 mM d'HEPES ;
    - 0,5 à 3 mM de proline, de préférence 1 mM de proline ; et
    - 5 500 à 7 000 mg/l de chlorure de sodium, de préférence 6 100 mg/l de chlorure de sodium,
    pour la culture d'une cellule CHO productrice d'interféron bêta humain recombinant dans un procédé comprenant une phase de croissance I, une phase de croissance II et une phase de production, la phase de croissance I étant effectuée à 37 °C, la phase de croissance II étant effectuée à 35 °C et la phase de production étant effectuée à 33 °C, et la phase de croissance II durant 1-2 jours.
EP05784517.4A 2005-08-26 2005-08-26 Procede de preparation d'interferon beta glycosyle Active EP1917276B1 (fr)

Priority Applications (6)

Application Number Priority Date Filing Date Title
RS20180470A RS57549B1 (sr) 2005-08-26 2005-08-26 Proces za pripremu glikoziliranog interferona beta
EP20110161155 EP2390263A1 (fr) 2005-08-26 2005-08-26 Procédé pour la préparation d'interféron bêta glycosylé
PT57845174T PT1917276T (pt) 2005-08-26 2005-08-26 Processo para a preparação de interferão beta glicosilado
PL05784517T PL1917276T3 (pl) 2005-08-26 2005-08-26 Sposób wytwarzania glikozylowanego interferonu beta
SI200532202T SI1917276T1 (en) 2005-08-26 2005-08-26 PROCEDURE FOR PREPARATION OF GLYCOSYLATED INTERFERONE BETA
HRP20180579TT HRP20180579T1 (hr) 2005-08-26 2018-04-11 Postupak za priređivanje glikoziliranog interferona beta

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/EP2005/054220 WO2007022799A1 (fr) 2005-08-26 2005-08-26 Procede de preparation d'interferon beta glycosyle

Related Child Applications (1)

Application Number Title Priority Date Filing Date
EP20110161155 Division-Into EP2390263A1 (fr) 2005-08-26 2005-08-26 Procédé pour la préparation d'interféron bêta glycosylé

Publications (2)

Publication Number Publication Date
EP1917276A1 EP1917276A1 (fr) 2008-05-07
EP1917276B1 true EP1917276B1 (fr) 2018-03-21

Family

ID=35788670

Family Applications (2)

Application Number Title Priority Date Filing Date
EP20110161155 Withdrawn EP2390263A1 (fr) 2005-08-26 2005-08-26 Procédé pour la préparation d'interféron bêta glycosylé
EP05784517.4A Active EP1917276B1 (fr) 2005-08-26 2005-08-26 Procede de preparation d'interferon beta glycosyle

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP20110161155 Withdrawn EP2390263A1 (fr) 2005-08-26 2005-08-26 Procédé pour la préparation d'interféron bêta glycosylé

Country Status (22)

Country Link
US (2) US20080219952A1 (fr)
EP (2) EP2390263A1 (fr)
JP (1) JP4944112B2 (fr)
KR (1) KR101276367B1 (fr)
CN (1) CN101282990B (fr)
AU (1) AU2005335900B2 (fr)
BR (1) BRPI0520498B8 (fr)
DK (1) DK1917276T3 (fr)
EA (1) EA015901B1 (fr)
ES (1) ES2664924T3 (fr)
HK (1) HK1120052A1 (fr)
HR (1) HRP20180579T1 (fr)
HU (1) HUE038947T2 (fr)
IL (1) IL189738A (fr)
LT (1) LT1917276T (fr)
MX (1) MX2008002596A (fr)
PL (1) PL1917276T3 (fr)
PT (1) PT1917276T (fr)
RS (1) RS57549B1 (fr)
SG (1) SG155183A1 (fr)
SI (1) SI1917276T1 (fr)
WO (1) WO2007022799A1 (fr)

Families Citing this family (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG155183A1 (en) 2005-08-26 2009-09-30 Ares Trading Sa Process for the preparation of glycosylated interferon beta
AU2006323925B2 (en) * 2005-12-09 2012-08-02 Ares Trading S.A. Method for purifying FSH or a FSH mutant
WO2008033517A2 (fr) 2006-09-13 2008-03-20 Abbott Laboratories Améliorations à la culture de cellules
US8911964B2 (en) 2006-09-13 2014-12-16 Abbvie Inc. Fed-batch method of making human anti-TNF-alpha antibody
EP2076533B1 (fr) 2007-05-02 2014-10-08 Ambrx, Inc. Polypeptides d'interferon beta modifies et leurs utilisations
JP2011500756A (ja) * 2007-10-22 2011-01-06 メルク セローノ ソシエテ アノニム Fc融合タンパク質を精製する方法
US20100256337A1 (en) * 2007-10-22 2010-10-07 Merck Serono Sa Method for purifying an fc-containing protein
DE102008051574A1 (de) * 2008-10-14 2010-04-15 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Verfahren zur Herstellung von Interferon-beta und deren Varianten
CN104974251A (zh) 2008-10-20 2015-10-14 Abbvie公司 在抗体纯化过程中的病毒灭活
EP2921501A1 (fr) * 2008-10-20 2015-09-23 Abbvie Inc. Isolement et purification d'anticorps par chromatographie d'affinité de la protéine A
WO2010089126A2 (fr) * 2009-02-09 2010-08-12 Roche Glycart Ag Analyse de motif de glycosylation d'immunoglobuline
DE102009032179A1 (de) * 2009-07-07 2011-01-13 Biogenerix Ag Verfahren zur Reinigung von Interferon beta
US20120177603A1 (en) * 2009-07-07 2012-07-12 Biogenerix Ag Method for the purification of interferon-b
US20110136682A1 (en) * 2009-12-04 2011-06-09 Momenta Pharmaceuticals, Inc. Antennary fucosylation in glycoproteins from cho cells
EP2702077A2 (fr) 2011-04-27 2014-03-05 AbbVie Inc. Procédé de contrôle du profil de galactosylation de protéines exprimées de manière recombinante
MY168902A (en) * 2011-10-01 2018-12-04 Glytech Inc Glycosylated polypeptide and pharmaceutical composition containing same
CN102533658B (zh) * 2011-12-08 2014-06-04 深圳新鹏生物工程有限公司 制备人干扰素β1a的细胞的培养方法和培养基及其应用
US9067990B2 (en) 2013-03-14 2015-06-30 Abbvie, Inc. Protein purification using displacement chromatography
WO2013158279A1 (fr) 2012-04-20 2013-10-24 Abbvie Inc. Procédés de purification de protéines pour réduire des espèces acides
WO2013158273A1 (fr) 2012-04-20 2013-10-24 Abbvie Inc. Procédés de modulation de la distribution de variant de lysine c-terminal
WO2013176754A1 (fr) 2012-05-24 2013-11-28 Abbvie Inc. Nouvelle purification d'anticorps au moyen de chromatographie à interaction hydrophobe
CN102757937B (zh) * 2012-07-05 2014-04-16 复旦大学附属中山医院 一种培养神经元的含酚红无血清培养体系以及酚红新用途
WO2014035475A1 (fr) 2012-09-02 2014-03-06 Abbvie Inc. Procédés de contrôle de l'hétérogénéité des protéines
US9512214B2 (en) 2012-09-02 2016-12-06 Abbvie, Inc. Methods to control protein heterogeneity
US9169331B2 (en) 2012-12-21 2015-10-27 Dionex Corporation Separation of glycans by mixed-mode liquid chromatography
US9310344B2 (en) 2013-06-14 2016-04-12 Dionex Corporation HILIC/anion-exchange/cation-exchange multimodal media
AU2013381687A1 (en) 2013-03-12 2015-09-24 Abbvie Inc. Human antibodies that bind human TNF-alpha and methods of preparing the same
WO2014159579A1 (fr) 2013-03-14 2014-10-02 Abbvie Inc. Anticorps anti-tnfα ayant mutés et leurs procédés d'utilisation
US9499614B2 (en) 2013-03-14 2016-11-22 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosaccharides
US9017687B1 (en) 2013-10-18 2015-04-28 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
US9598667B2 (en) 2013-10-04 2017-03-21 Abbvie Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
LT6164B (lt) 2013-10-15 2015-06-25 Uab Biotechnologinės Farmacijos Centras "Biotechpharma" Sulieti interferono-alfa 5 baltymai su kitu citokinu ir jų gamybos būdas
US9181337B2 (en) 2013-10-18 2015-11-10 Abbvie, Inc. Modulated lysine variant species compositions and methods for producing and using the same
US9085618B2 (en) 2013-10-18 2015-07-21 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US8946395B1 (en) 2013-10-18 2015-02-03 Abbvie Inc. Purification of proteins using hydrophobic interaction chromatography
US20150139988A1 (en) 2013-11-15 2015-05-21 Abbvie, Inc. Glycoengineered binding protein compositions
AU2015239104B2 (en) * 2014-04-04 2018-12-20 Ares Trading S.A. Novel IFN beta protein analogs
EA027609B9 (ru) * 2015-04-29 2017-11-30 Тютор С.А.С.И.Ф.И.А. Способ получения интерферона бета-1альфа и фармацевтическая композиция, содержащая интерферон бета-1альфа
EP3303559A1 (fr) * 2015-06-01 2018-04-11 Biogen MA Inc. Supplémentation en manganèse pour la régulation de la glycosylation dans un procédé de culture cellulaire de mammifère
KR20210009982A (ko) * 2019-07-18 2021-01-27 에이비온 주식회사 2당화된 인터페론-베타 단백질의 정제 방법
CN111563198B (zh) * 2020-04-16 2023-07-25 百度在线网络技术(北京)有限公司 一种物料召回方法、装置、设备及存储介质
CN117024561B (zh) * 2023-10-10 2024-02-20 哈药集团生物工程有限公司 一种聚乙二醇修饰干扰素的纯化方法

Family Cites Families (75)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5326859A (en) 1979-10-30 1994-07-05 Juridical Foundation, Japanese Foundation For Cancer Research DNA and recombinant plasmid
CH648331A5 (de) 1979-07-31 1985-03-15 Hoffmann La Roche Homogenes fibroblasten-interferon und dessen herstellung.
DE3273787D1 (en) 1981-02-04 1986-11-20 Japan Found Cancer Human interferon-beta gene
US4588585A (en) 1982-10-19 1986-05-13 Cetus Corporation Human recombinant cysteine depleted interferon-β muteins
US4737462A (en) 1982-10-19 1988-04-12 Cetus Corporation Structural genes, plasmids and transformed cells for producing cysteine depleted muteins of interferon-β
US4966843A (en) * 1982-11-01 1990-10-30 Cetus Corporation Expression of interferon genes in Chinese hamster ovary cells
US4666828A (en) 1984-08-15 1987-05-19 The General Hospital Corporation Test for Huntington's disease
US4959314A (en) 1984-11-09 1990-09-25 Cetus Corporation Cysteine-depleted muteins of biologically active proteins
US5116943A (en) 1985-01-18 1992-05-26 Cetus Corporation Oxidation-resistant muteins of Il-2 and other protein
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US4801531A (en) 1985-04-17 1989-01-31 Biotechnology Research Partners, Ltd. Apo AI/CIII genomic polymorphisms predictive of atherosclerosis
US5266476A (en) 1985-06-18 1993-11-30 Yeda Research & Development Co., Ltd. Fibrous matrix for in vitro cell cultivation
US5017691A (en) 1986-07-03 1991-05-21 Schering Corporation Mammalian interleukin-4
US4879111A (en) 1986-04-17 1989-11-07 Cetus Corporation Treatment of infections with lymphokines
US5045468A (en) 1986-12-12 1991-09-03 Cell Enterprises, Inc. Protein-free culture medium which promotes hybridoma growth
NO162160C (no) 1987-01-09 1989-11-15 Medi Cult As Serumfritt vekstmedium, samt anvendelse derav.
DE3712564A1 (de) 1987-04-14 1988-11-24 Bioferon Biochem Substanz Verfahren zur konstruktion einer animalen zellinie fuer die herstellung von humanem interferon-beta
IT1206302B (it) 1987-06-26 1989-04-14 Serono Cesare Ist Ricerca Ormone follicolo-stimolante urinario
US4965195A (en) 1987-10-26 1990-10-23 Immunex Corp. Interleukin-7
US4904584A (en) 1987-12-23 1990-02-27 Genetics Institute, Inc. Site-specific homogeneous modification of polypeptides
US6048728A (en) 1988-09-23 2000-04-11 Chiron Corporation Cell culture medium for enhanced cell growth, culture longevity, and product expression
US5272057A (en) 1988-10-14 1993-12-21 Georgetown University Method of detecting a predisposition to cancer by the use of restriction fragment length polymorphism of the gene for human poly (ADP-ribose) polymerase
US5338835A (en) * 1989-02-21 1994-08-16 Washington University CTP-extended form of FSH
WO1990009800A1 (fr) * 1989-02-21 1990-09-07 Washington University Formes modifiees d'hormones de reproduction
US5192659A (en) 1989-08-25 1993-03-09 Genetype Ag Intron sequence analysis method for detection of adjacent and remote locus alleles as haplotypes
US5665868A (en) 1990-09-14 1997-09-09 Vittal Mallya Scientific Research Foundation Chromatographic agent and its use for the separation or proteins, polypeptides of metals
RU2166955C2 (ru) 1991-01-18 2001-05-20 Эмген Инк. Способ лечения и профилактики заболеваний, обусловленных повышением уровня фактора, вызывающего некроз опухолевых клеток
US5508261A (en) * 1991-06-18 1996-04-16 University Of Medicine & Dentistry Of New Jersey Analogs of glycoprotein hormones having altered receptor binding specificity and activity and methods for preparing and using same
DE4128319A1 (de) 1991-08-27 1993-03-04 Bioferon Biochem Substanz Neues rekombinantes human-ifn-beta, verfahren zu dessen herstellung und dieses enthaltende pharmazeutische zubereitungen
SE9201073D0 (sv) * 1992-04-03 1992-04-03 Kabi Pharmacia Ab Protein formulation
ATE501254T1 (de) 1992-10-23 2011-03-15 Genetics Inst Llc P- selectin ligandenprotein
EP0666312A1 (fr) 1994-02-08 1995-08-09 Wolfgang A. Renner Procédé pour l'amélioration de la croissance de cellules de mammifères
US5545723A (en) * 1994-03-15 1996-08-13 Biogen Inc. Muteins of IFN-β
AU720337B2 (en) 1995-02-17 2000-05-25 Stichting Dienst Landbouwkundig Onderzoek Production of biologically active recombinant bovine follicle stimulating hormone (REC bFSH) in the baculovirus expression system
AUPN801296A0 (en) 1996-02-12 1996-03-07 Csl Limited Stabilised growth hormone formulation and method of preparation thereof
DE69623109T2 (de) 1996-04-19 2002-12-12 Nestle Sa Menschliche immortalisierte Colon-Epithelialzellen
JP2000517188A (ja) * 1996-08-30 2000-12-26 ライフ テクノロジーズ,インコーポレイテッド 無血清哺乳動物細胞培養培地およびその使用
IT1287138B1 (it) 1996-11-07 1998-08-04 Ibsa Inst Biochimique Sa Procedimento per la separazione e purificazione di fsh e lh
US6162905A (en) * 1996-11-07 2000-12-19 Ibsa Institut Biochimique S.A. FSH and LH separation and purification process
CZ300296B6 (cs) 1996-11-15 2009-04-15 Genentech, Inc. Zpusob izolace rekombinantního lidského neurotrofinu, neurotrofinový prostredek a zpusob cištení neurotrofinu
US5883073A (en) * 1997-04-03 1999-03-16 Washington University Single-chain double-alpha peptide
US6013253A (en) 1997-08-15 2000-01-11 Amgen, Inc. Treatment of multiple sclerosis using consensus interferon and IL-1 receptor antagonist
EP1224940B2 (fr) 1997-09-23 2010-08-11 Rentschler Biotechnologie GmbH Formulations liquides d' interféron-bèta
US6414123B1 (en) 1997-10-21 2002-07-02 Vitro Diagnostics, Inc. Method for purifying FSH
US5990288A (en) * 1997-10-21 1999-11-23 Vitro Diagnostics, Inc. Method for purifying FSH
EA003789B1 (ru) 1998-04-28 2003-10-30 Апплайд Резеч Системз Арс Холдинг Н.В. КОНЪЮГАТЫ ПОЛИОЛ-β-ИНТЕРФЕРОН
US20030153042A1 (en) 1998-07-28 2003-08-14 California Institute Of Technology Expression of functional eukaryotic proteins
ATE358139T1 (de) 1999-04-16 2007-04-15 Inst Massone S A Verfahren zur herstelllung von gonadotropin- zusammensetzungen
US7431921B2 (en) * 2000-04-14 2008-10-07 Maxygen Aps Interferon beta-like molecules
WO2001058493A1 (fr) 2000-02-11 2001-08-16 Maxygen Aps Conjugates of follicle stimulating hormones
RS50416B (sr) 2000-02-22 2009-12-31 Laboratoires Serono Sa., Prečišćeni lh
JP2003530838A (ja) * 2000-04-12 2003-10-21 ヒューマン ゲノム サイエンシズ インコーポレイテッド アルブミン融合タンパク質
US20030017550A1 (en) * 2001-01-22 2003-01-23 Pang Danny Zhong Der DNA sequences encoding fusion proteins comprising IFN-beta and TM-alpha1
HUP0303251A2 (hu) * 2001-02-27 2003-12-29 Maxygen Aps Béta-interferon-szerű molekulák
US20030096414A1 (en) 2001-03-27 2003-05-22 Invitrogen Corporation Culture medium for cell growth and transfection
AU784808B2 (en) 2001-04-02 2006-06-29 Kedrion Melville Inc. Prion and viral clearance process
CN1522159A (zh) * 2001-06-29 2004-08-18 马克西根公司 干扰素配制品
US7226903B2 (en) * 2001-10-10 2007-06-05 Neose Technologies, Inc. Interferon beta: remodeling and glycoconjugation of interferon beta
US7317095B2 (en) 2001-10-22 2008-01-08 Applied Research Systems Ars Holding N.V. Mutant glycoproteins
CA2471363C (fr) * 2001-12-21 2014-02-11 Human Genome Sciences, Inc. Proteines hybrides d'albumine
JP2005519946A (ja) * 2002-03-12 2005-07-07 マキシジェン エーピーエス 発作の処置のためのインターフェロンβ様分子
CN101664545A (zh) 2002-07-17 2010-03-10 比奥根艾迪克Ma公司 使用干扰素-β治疗肾衰竭
TWI353991B (en) 2003-05-06 2011-12-11 Syntonix Pharmaceuticals Inc Immunoglobulin chimeric monomer-dimer hybrids
UA88879C2 (en) 2003-09-02 2009-12-10 Эплайд Рисерч Системз Эрс Холдинг Н.В. Fsh glycosylation mutant
KR100524871B1 (ko) * 2003-12-04 2005-10-31 씨제이 주식회사 인터페론 베타의 정제방법
KR100524872B1 (ko) * 2003-12-04 2005-11-01 씨제이 주식회사 인터페론 베타의 정제방법
JP4763616B2 (ja) * 2003-12-22 2011-08-31 アレス トレーディング ソシエテ アノニム Fshを精製するための方法
US7598356B2 (en) 2004-07-08 2009-10-06 Board of Regents of the University of Nebraska by and on behalf of the University of Nebraska Medical Center Method for purifying a protein of the cystine-knot superfamily
TWI384069B (zh) * 2004-08-27 2013-02-01 Pfizer Ireland Pharmaceuticals 多胜肽之製法
MX2007005327A (es) * 2004-11-09 2007-08-02 Ares Trading Sa Metodo para purificacion de la hormona foliculo estimulante (hfe).
SG155183A1 (en) 2005-08-26 2009-09-30 Ares Trading Sa Process for the preparation of glycosylated interferon beta
AU2006323925B2 (en) * 2005-12-09 2012-08-02 Ares Trading S.A. Method for purifying FSH or a FSH mutant
US7956034B2 (en) * 2006-01-17 2011-06-07 Merck Serono Sa FSH glycosylation variant D3N
JP2011500756A (ja) * 2007-10-22 2011-01-06 メルク セローノ ソシエテ アノニム Fc融合タンパク質を精製する方法
US20100256337A1 (en) * 2007-10-22 2010-10-07 Merck Serono Sa Method for purifying an fc-containing protein

Also Published As

Publication number Publication date
WO2007022799A1 (fr) 2007-03-01
AU2005335900A1 (en) 2007-03-01
IL189738A (en) 2012-07-31
JP2009505645A (ja) 2009-02-12
CN101282990A (zh) 2008-10-08
IL189738A0 (en) 2008-08-07
HUE038947T2 (hu) 2018-12-28
BRPI0520498A2 (pt) 2010-07-13
EA015901B1 (ru) 2011-12-30
EP2390263A1 (fr) 2011-11-30
AU2005335900B2 (en) 2012-03-29
EA200800669A1 (ru) 2008-12-30
PT1917276T (pt) 2018-06-11
US8993724B2 (en) 2015-03-31
MX2008002596A (es) 2008-03-14
KR101276367B1 (ko) 2013-06-25
SI1917276T1 (en) 2018-05-31
DK1917276T3 (en) 2018-05-07
HRP20180579T1 (hr) 2018-05-18
ES2664924T3 (es) 2018-04-24
BRPI0520498B8 (pt) 2021-05-25
CN101282990B (zh) 2013-04-03
JP4944112B2 (ja) 2012-05-30
KR20080048490A (ko) 2008-06-02
HK1120052A1 (en) 2009-03-20
LT1917276T (lt) 2018-05-25
RS57549B1 (sr) 2018-10-31
EP1917276A1 (fr) 2008-05-07
SG155183A1 (en) 2009-09-30
PL1917276T3 (pl) 2018-08-31
US20110305669A1 (en) 2011-12-15
US20080219952A1 (en) 2008-09-11
BRPI0520498B1 (pt) 2018-11-06

Similar Documents

Publication Publication Date Title
EP1917276B1 (fr) Procede de preparation d'interferon beta glycosyle
O’Flaherty et al. Mammalian cell culture for production of recombinant proteins: A review of the critical steps in their biomanufacturing
EP1720979B1 (fr) Utilisation d'un milieu de culture de cellules exempt de serum et destine a la production de il-18bp dans des cellules mammiferes
CA2234215C (fr) Preparation du facteur recombinant viii dans un milieu exempt de proteines
Schäffer et al. Nitric oxide, an autocrine regulator of wound fibroblast synthetic function.
Bragonzi et al. A new Chinese hamster ovary cell line expressing α2, 6-sialyltransferase used as universal host for the production of human-like sialylated recombinant glycoproteins
Todd-Turla et al. CFTR expression in cortical collecting duct cells
US20060177932A1 (en) Expansion agents for stem cells
EP1877545A2 (fr) Systemes d'expression chez le mammifere
Gu et al. Influence of Primatone RL supplementation on sialylation of recombinant human interferon‐γ produced by Chinese hamster ovary cell culture using serum‐free media
US20080038824A1 (en) Composition and Method for Elevating Gene Transfer Efficiency
Kyburz et al. Effects of methotrexate on thymidine triphosphate levels in Chinese hamster cell cultures
CA2096329A1 (fr) Methode pour accroitre et activer les monocytes et les polynucleaires neutrophiles et pour favoriser la maturation des cellules myeloides a l'aide de l'interleukine-5
JPS59140898A (ja) インタ−ロイキン−2の製造法

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20080215

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR MK YU

17Q First examination report despatched

Effective date: 20090406

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20171009

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR MK YU

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: MERCK SERONO S.A. INTELLECTUAL PROPERTY, CH

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: HR

Ref legal event code: TUEP

Ref document number: P20180579

Country of ref document: HR

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 980955

Country of ref document: AT

Kind code of ref document: T

Effective date: 20180415

REG Reference to a national code

Ref country code: RO

Ref legal event code: EPE

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602005053698

Country of ref document: DE

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2664924

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20180424

REG Reference to a national code

Ref country code: NL

Ref legal event code: FP

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

Effective date: 20180503

REG Reference to a national code

Ref country code: HR

Ref legal event code: T1PR

Ref document number: P20180579

Country of ref document: HR

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: PT

Ref legal event code: SC4A

Ref document number: 1917276

Country of ref document: PT

Date of ref document: 20180611

Kind code of ref document: T

Free format text: AVAILABILITY OF NATIONAL TRANSLATION

Effective date: 20180604

REG Reference to a national code

Ref country code: CH

Ref legal event code: PCAR

Free format text: NEW ADDRESS: TERRE BONNE BUSINESS PARK A2 ROUTE DE CRASSIER 15, 1262 EYSINS (CH)

Ref country code: EE

Ref legal event code: FG4A

Ref document number: E015360

Country of ref document: EE

Effective date: 20180418

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 14

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180321

REG Reference to a national code

Ref country code: SK

Ref legal event code: T3

Ref document number: E 27093

Country of ref document: SK

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180622

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180621

REG Reference to a national code

Ref country code: HU

Ref legal event code: AG4A

Ref document number: E038947

Country of ref document: HU

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602005053698

Country of ref document: DE

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20190102

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180321

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20180579

Country of ref document: HR

Payment date: 20190715

Year of fee payment: 15

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20180579

Country of ref document: HR

Payment date: 20200807

Year of fee payment: 16

REG Reference to a national code

Ref country code: AT

Ref legal event code: UEP

Ref document number: 980955

Country of ref document: AT

Kind code of ref document: T

Effective date: 20180321

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20180579

Country of ref document: HR

Payment date: 20210820

Year of fee payment: 17

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20180579

Country of ref document: HR

Payment date: 20220823

Year of fee payment: 18

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230518

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: PT

Payment date: 20230630

Year of fee payment: 19

Ref country code: DK

Payment date: 20230627

Year of fee payment: 19

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: PL

Payment date: 20230616

Year of fee payment: 19

Ref country code: NL

Payment date: 20230718

Year of fee payment: 19

Ref country code: LV

Payment date: 20230621

Year of fee payment: 19

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20180579

Country of ref document: HR

Payment date: 20230721

Year of fee payment: 19

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: LU

Payment date: 20230814

Year of fee payment: 19

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: TR

Payment date: 20230823

Year of fee payment: 19

Ref country code: RO

Payment date: 20230712

Year of fee payment: 19

Ref country code: IT

Payment date: 20230711

Year of fee payment: 19

Ref country code: IE

Payment date: 20230710

Year of fee payment: 19

Ref country code: GB

Payment date: 20230706

Year of fee payment: 19

Ref country code: FI

Payment date: 20230816

Year of fee payment: 19

Ref country code: ES

Payment date: 20230901

Year of fee payment: 19

Ref country code: EE

Payment date: 20230719

Year of fee payment: 19

Ref country code: CZ

Payment date: 20230817

Year of fee payment: 19

Ref country code: CH

Payment date: 20230902

Year of fee payment: 19

Ref country code: AT

Payment date: 20230725

Year of fee payment: 19

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: SK

Payment date: 20230712

Year of fee payment: 19

Ref country code: SI

Payment date: 20230627

Year of fee payment: 19

Ref country code: SE

Payment date: 20230630

Year of fee payment: 19

Ref country code: IS

Payment date: 20230711

Year of fee payment: 19

Ref country code: HU

Payment date: 20230720

Year of fee payment: 19

Ref country code: FR

Payment date: 20230703

Year of fee payment: 19

Ref country code: DE

Payment date: 20230703

Year of fee payment: 19

Ref country code: BE

Payment date: 20230719

Year of fee payment: 19

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: LT

Payment date: 20230808

Year of fee payment: 19